Efficacy and safety of convalescent plasma to treat hospitalised COVID-19 patients with or without underlying immunodeficiency: a randomized clinical trial =========================================================================================================================================================== * Karine Lacombe * Thomas Hueso * Raphael Porcher * Arsène Mekinian * Thibault Chiarabini * Sophie Georgin-Lavialle * Florence Ader * Julien Saison * Guillaume Martin Blondel * Nathalie De Castro * Fabrice Bonnet * Charles Cazanave * Anne François * Pascal Morel * Olivier Hermine * Valérie Pourcher * Marc Michel * Xavier Lescure * Nora Soussi * Philippe Brun * Fanny Pommeret * Pierre-Olivier Sellier * Stella Rousset * Lionel Piroth * Jean-Marie Michot * Gabriel Baron * Xavier De Lamballerie * Xavier Mariette * Pierre-Louis Tharaux * Matthieu Resche-Rigon * Philippe Ravaud * Tabassome Simon * Pierre Tiberghien ## ABSTRACT **Background** Efficacy of COVID-19 convalescent plasma (CCP) in COVID-19 pneumonia is uncertain. Early transfusion of high antibody titre CCP may be beneficial, especially in case of underlying immunodeficiency. **Methods** The CORIPLASM study was a multicentric, open-label, Bayesian randomised clinical trial evaluating the efficacy of CCP in patients with moderate COVID-19 pneumonia, including patients with underlying immunosuppression. Patients hospitalised with COVID-19 for less than 9 days were assigned to receive 2 plasma units/day over 2 days (CCP) or usual care (UC) alone. Primary outcomes were the proportion of patients with a WHO-Clinical Progression Score (CPS) ≥6 on the 10-point scale on day (d) 4 and survival without ventilation or additional immunomodulatory treatment by d14. Main analysis was conducted on the whole population and a planned subgroup analysis was performed according to immunosuppression status. **Findings** A total of 120 patients were recruited between April 16, 2020, and April 21, 2021, and assigned to CCP (n=60) or UC (n=60) with a 28 day-follow-up. The median time from symptoms onset to randomisation (days) was 7.0 [interquartile range (IQR) 5.0-9.0] and 7.0 [IQR 4.0-8.5] in CCP and UC, respectively. Thirteen (22%) patients with CCP had a WHO-CPS ≥6 at d4 versus 8 (13%) with UC, adjusted odds ratio (aOR) 1.88 [95% confidence interval (CI), 0.71 to 5.24]. By d14, 19 (31.6%) patients with CCP and 20 (33.3%) patients with UC had ventilation, additional immunomodulatory treatment or had died. Cumulative incidence of death was 3 (5%) with CCP and 8 (13%) with UC at d14 (aHR 0.40 [95%CI 0·10 -1·53]), and 7 (12%) with CCP and 12 (20%) with UC at d28 (aHR 0.51 [95% CI 0.20-1.32]). Subgroup analysis indicated that CCP might be associated with a lower mortality in patients with underlying immunosuppression (HR 0.37 [95% CI 0.14-0.97]). Serious adverse events were noted in 30 (50%) and 26 (43%) patients with CCP or UC, respectively. **Interpretation** CCP treatment did not improve early outcomes in patients with mild-to-moderate form COVID-19 pneumonia but was associated with reduced mortality in the subgroup of immunosuppressed patients. **Trial registration** [clinicaltrials.gov](http://clinicaltrials.gov) Identifier: [NCT04345991](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04345991&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) **Funding** Programme Hospitalier de Recherche Clinique / DGOS; Fondation pour la Recherche Médicale ; Sorbonne Université Paris; Emergency support instrument, DG Santé, European Commission ## INTRODUCTION Early in the COVID-19 pandemic, COVID-19 convalescent plasma (CCP) transfusion was identified as a potential treatment that needed evaluation.1 Overall efficacy of CCP in hospitalized patients has not been established.2 However, high titre CCP may be beneficial particularly if used early before seroconversion3,4 or in patients unable to mount an efficient humoral response.5,6 Monoclonal antibody treatment has demonstrated efficacy as an early intervention7 or later in hospitalised seronegative patients,8 however with significant limitations including accessibility and cost,9 as well as a risk of efficacy loss as recently exemplified with the emergence of the delta and omicron SARS-CoV-2 variants.10,11 By contrast, CCP from convalescent vaccinated donors is cheaper, readily available and adaptable to a changing viral landscape, and potentially less prone to immune resistance. While the recent omicron waves have been associated with a steep decrease in the efficacy of almost all available monoclonal antibodies, high titre CCP from (pre-omicron) convalescent vaccinated donors may retain anti-omicron neutralization activity.12 Such anti-micron neutralisation capacity is further increased in CCP from omicron convalescent vaccinated donors.13 Oral direct antiviral agents such as molnupiravir14 or nirmatrelvir/ritonavir15 also represent another therapeutic option, but they have drawbacks, such as necessitating an initiation within 5 days of symptoms onset and drug interactions for nirmatrelvir/ritonavir, notably in immunosuppressed patients (in whom data on efficacy and safety of oral treatment are scant). Furthermore, these oral antivirals are not indicated in patients requiring oxygen. Lastly, the intravenous antiviral agent remdesivir has demonstrated only limited anti-SARS-CoV-2 efficacy in hospitalised patients.16 Careful assessment of CCP efficacy and safety remains an important public health issue, particularly in immunosuppressed patients unable to mount a vaccine-mediated immune response and at risk of severe disease with limited therapeutic options. We report the results of a randomised controlled trial assessing the efficacy of CCP (4 units, ≈ 850 ml) in immune-competent and immunosuppressed patients hospitalized for moderate SARS-CoV-2 associated pneumonia requiring no assisted ventilation at time of inclusion. ## METHODS ### Trial design and study oversight CORIMUNO-19 is a platform trial established by Assistance Publique-Hôpitaux de Paris, France, at the early beginning of the COVID-19 pandemic.17 CORIMUNO-CORIPLASM was an embedded multicentric, open-label randomized controlled trial in patients with moderate COVID-19 pneumonia conducted in 14 French hospitals across France ([NCT04345991](http://medrxiv.org/lookup/external-ref?link_type=CLINTRIALGOV&access_num=NCT04345991&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom)). Ethical clearance was obtained from CPP Ile de France VI on April 10, 2020 (no. 26-20 Med.1°). ### Study population and randomization At hospital admission, patients were evaluated for eligibility criteria: hospitalized adult ≥18 years of age, positive SARS-CoV-2 nasopharyngeal PCR and/or CT scan prior to randomisation, onset of symptoms <9 days, illness of mild or moderate severity according to the WHO clinical progression scale (CPS) (hospitalised, mild disease: no oxygen need; hospitalised, moderate disease: oxygen need <3l; (Appendix), no pregnancy, no prior severe grade 3 allergic reaction to plasma transfusion, and no current documented bacterial infection. ABO compatibility with available CCP was verified before patient inclusion. Written informed consent was obtained from all patients or their legal representatives at inclusion in CORIMUNO19. A specific written informed consent was sought from eligible patients before inclusion in the CORIPLASM trial. The independent clinical research organisation drew up the computerized randomization list, and the patient’s randomization number was accessed through a secure site by a site study team member. Randomisation was performed within 2 hours after enrolment. Eligible patients were randomised 1:1 to receive either convalescent plasma or usual care. The latter could include the use of dexamethasone, tocilizumab, supportive care including supplemental oxygen, antivirals, and antibiotics. A Data and Safety Monitoring Board (DSMB) provided guidance to the trial after inclusion of every 60 patients. ### Study product Convalescent donors were eligible for plasma donation 15 days after the resolution of COVID-19-related symptoms. Collected apheresis plasma by Etablissement Français du Sang (EFS) underwent pathogen reduction (INTERCEPT Blood System, Cerus, Concord, CA) and standard testing as per current regulations in France. Anti-SARS-CoV-2 potency was assessed in each donation, with a requirement for a SARS-CoV-2 seroneutralization titer ≥=40.18 Additionally, antibody content was determined by IgG enzyme-linked immunosorbent assay (ELISA EUROIMMUN, Bussy-Saint-Martin, France). CCP made available for the trial, all collected between April and June 2020, yielded a mean ELISA ratio of 6.1 (s.d.: 2.9). After the first 3 patients received 2 units of ABO-compatible CCP as per protocol, all subsequent patients randomised to the CCP arm received 4 units of CCP (200-220 ml/unit, 2 units/day over 2 consecutive days) provided by different donors. ### Study endpoints As in all CORIMUNO19 nested trials, there was an early primary endpoint defined as a WHO-CPS ≥ 6 (Supp. material, Appendix) at day (d)4 of randomization. The primary endpoint specific to the CORIPLASM trial was survival without the need for ventilator use (including non-invasive ventilation, NIV or high flow oxygen) at d14 of randomization (WHO-CPS < 6) or additional immunomodulatory treatment except corticosteroids. Secondary endpoints included WHO-CPS at 4, 7 and 14 days after randomization, overall survival at 14 and 28 days after randomization, time to discharge, time to oxygen supply independency and evolution of a series of biological parameters at days 4, 7 and 14 after randomization. Pre-defined subgroups analyses included immunosuppression status (underlying immunodeficiency: yes/no), duration of symptoms before randomization (≤5 days, > 5 days), and use of steroids. Safety data included all clinical and biological adverse events observed during study follow-up. Immunodeficiency was defined as the presence of at least one of the following medical conditions: active malignant neoplasm, lymphoid or myeloid neoplasms, hematopoietic stem cell or solid organ transplantation, or HIV/AIDS not on highly active antiretroviral treatment. ### Statistical Analysis The sample size was set at 120 participants (60 per group), with a Bayesian interim analysis after 60 participants were randomised. We computed that the trial would have a frequentist power of 97.2% to detect a decrease in event rate from 0.50 to 0.20, and 73.9% to detect a decrease in event rates from 0.50 to 0.30. The study statisticians, who were masked to the group assignment, oversaw the interim and final analyses. Interim analysis reports were only shared with DSMB members and not with trial investigators, who remained blinded to all results during the trial. The treatment effect was primarily expressed as an absolute risk difference (ARD) for the early primary endpoint, and a hazard ratio (HR) for the longer-term primary endpoint. Both were analysed in a Bayesian framework. A posterior probability of ARD <0 or HR <1 greater than 0.99 at the interim analysis or greater than 0.95 at the final analysis indicated efficacy. We also computed posterior probabilities of ARD <-5.5% and HR <0.85, denoting a moderate or greater effect. At the interim analysis, a posterior probability of moderate or greater impact <0.20 defined a futility boundary. The treatment effect was summarised by the posterior median and equal tail credible intervals (CrIs). Because the decision rules are one-sided, consistent CrIs would theoretically be one-sided 95% CrIs, but we chose to report two-sided 90% CrIs with the same upper bound. For the early primary endpoint, the posterior distribution of ARD was computed analytically, using a beta prior distribution with parameters 1 and 1 for the proportion in each group. An odds ratio (OR) adjusted for age and centre was also estimated using a Bayesian logistic regression model. For the longer-term primary endpoint, the posterior HR distribution adjusted for age and centre was computed using Markov chain Monte Carlo (MCMC) with Gaussian prior distributions with mean 0 and variance 106 for the log HR. Different prior distributions were used as sensitivity analyses. Secondary outcomes were analysed in a frequentist framework, except for WHO-CPS scores, analysed as an ordinal variable with a Bayesian proportional odds model. Analyses were done on an intention-to-treat (ITT) basis. The original protocol specified a modified ITT analysis excluding patients declining the intervention and those unable to receive planned plasma therapy due to unavailability of ABO-compatible CCP. Since those situations did not occur, no modified ITT analysis was performed. No correction for multiplicity was done for secondary outcomes, and corresponding results should be regarded as exploratory. Two interim analyses were conducted (Table S1). Statistical analyses used SAS (version 9.4, SAS Institute) and R (version 4.0.5, R Foundation) statistical software. ## RESULTS ### Study population and CCP administration Between April 16, 2020, and April 21, 2021, a total of 120 patients (60 with CCP, 60 with UC only) were enrolled (Figure S1). Subjects’ characteristics are reported in Table 1. In both groups, the median time between the onset of symptoms and CCP transfusion was 7 days. A positive anti-S and anti-N SARS-CoV-2 serology was noted in 10/23 (44%) evaluable patients receiving CCP and 9/27 (33%) evaluable patients receiving UC. Thirty/sixty (50%) and 26/60 (44%) patients had an underlying immunodeficiency in the CCP and UC arms, respectively. One patient was considered to have COVID-19 with a typical chest CT scan at inclusion, but was then reclassified as non-indicative of SARS-CoV-2 infection and finally diagnosed with pulmonary oedema from cardiac origin. The other treatments received before and after randomisation until day 14 are reported in Table S2. View this table: [Table 1.](http://medrxiv.org/content/early/2022/08/10/2022.08.09.22278329/T1) Table 1. Characteristics at baseline. Values are median [interquartile range] unless stated otherwise. Intention to treat analysis was performed on 120 patients, of whom 2 in each study group were lost to follow-up at day 28 evaluation but discharged alive before day 28 (Figure 1). One patient did not receive any plasma infusion because of sudden worsening after randomisation and transfer to an intensive care unit (ICU), 9 patients received 2 units (3 as per protocol, 6 because of worsening of clinical status leading to ICU admission), and 50 received 4 units. Same day transfusion occurred in 78% of patients, whereas 12 (20%) and 1 (2%) were transfused 1 and 3 days after randomisation, respectively. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/08/10/2022.08.09.22278329/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2022/08/10/2022.08.09.22278329/F1) Figure 1: Flow chart of the CORIPLASM randomized clinical trial * No SARS-CoV-2 infection; † Under non-invasive ventilation. ### Primary outcomes Thirteen (22 %) patients in the CCP arm versus 8 (13%) patients in the UC arm had a WHO-CPS ≥6 at d4 (median posterior absolute risk difference +8·0%; 90% credible interval [CrI] –3.2-+19.4), adjusted odds ratio (aOR) 1.88 [95% CrI 0.71-5.24] (Tables 2 and S3, Figure S2). View this table: [Table 2.](http://medrxiv.org/content/early/2022/08/10/2022.08.09.22278329/T2) Table 2. Primary and secondary efficacy outcomes. By d14, 19 (31.6%) and 20 (33.3%) patients in the CCP and UC arms, respectively, needed non-invasive or high flow ventilation (CCP:15, UC:13) or additional immunomodulatory treatment (CCP:0, UC:5) or had died (2 in each arm, in addition to 1 and 5 deaths that occurred after reaching the primary outcome in the CCP and UC arm, respectively). The cumulative incidence of ventilation or death is reported in Figure 2a. The median posterior adjusted hazard ratio was 1.04 [95% CrI 0.55-1.97] (Tables 2 and S4, figure S3). ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/08/10/2022.08.09.22278329/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2022/08/10/2022.08.09.22278329/F2) Figure 2: 2a. Cumulative incidence of mechanical ventilation or death over 14 days (events at day 1 of randomisation occurred on the same day, but after randomisation); 2b. Cumulative incidence of death during follow-up; 2c.Distribution of WHO-CPS during follow-up. ### Secondary outcomes At d14, the cumulative incidence of death was 3 (5%) and 8 (13%) in the CCP and UC arms, respectively (aHR 0·40 [95% confidence interval (CI) 0.10-1.53])(Figure 2b, Table S5). At d28, 7 (12%) and 12 (20%) patients had died in the CCP and UC groups, respectively, with aHR 0.51 [95% CI 0.20-1.32]. The distribution of the WHO-CPS from day 1 to day 14 did not differ within groups, with a trend in the CCP group towards higher WHO-CPS between day 3 to 5 and lower mortality at day 14 (figure 2c, Table S6). At d14 and d28, 38 and 48 patients in the CCP group and 36 and 45 in the UC group were discharged, respectively, with an adjusted d28 sub distribution hazard ratio (SHR) of 0.99 [95% CI 0.65-1.49] adjusted for age and centre sub-distribution (Table S7). The incidence of oxygen supply independency until d28 was not different between groups: 76% and 62% by d14, 82% and 71% by d28, in the CCP and UC arms, respectively (SHR: 1.18, [95% CI 0.73-1.91])(Table S7). ### Subgroup analyses Figure 3 reports the primary outcome at d14, with no difference in the different subgroups. In the 49 patients who had an underlying immunodeficiency, the ratio of a WHO-CPS ≥6 at d4 was not statistically different in the CCP arm compared to the UC arm (27% versus 15%, aOR: 2.20 [95% CI 0.52-9.30]). At d28, there was a significantly better survival of immunosuppressed patients who received CCP compared to those with UC: 4/22 patients had died in the CCP group versus 11/27 in the UC group (HR 0.36, [95% CI 0.14-0.97])(Figure 4). Neither the symptoms duration nor the use of dexamethasone had an impact on day 28 survival. Post-hoc analysis of Ab potency in transfused CCP in relation to outcome did not reveal a significant dose-effect (Table S8). ![Figure 3:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/08/10/2022.08.09.22278329/F3.medium.gif) [Figure 3:](http://medrxiv.org/content/early/2022/08/10/2022.08.09.22278329/F3) Figure 3: Subgroup analysis of the d14 primary outcomes (death, need for non-invasive or mechanical ventilation, use of immunomodulatory drugs). The grey line indicates the overall estimate of treatment effect. Given only one patient was receiving antivirals at randomisation, no subgroup analysis according to antivirals was performed ![Figure 4:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2022/08/10/2022.08.09.22278329/F4.medium.gif) [Figure 4:](http://medrxiv.org/content/early/2022/08/10/2022.08.09.22278329/F4) Figure 4: Cumulative incidence of death during follow-up in patients with (4a) or without (4b) underlying immunodeficiency ### Safety Adverse events were reported in 44 (73%) and 36 (60%) patients in the CCP (n=124) and UC (n=103) arms, respectively: incidence rate ratio = 1.06 [95% CI 0.63-1.77])(Table S9). Serious adverse events were noted in 30 (50%) and 26 (43%) patients in the CCP (n=46) and UC (n=48) arms, respectively (incidence rate ratio = 0.84 [95%CI 0.46-1.54]). Of note, 10 sepsis-related events were observed with UC (6 with CCP) and 4 acute pulmonary oedema were reported with CCP (none with UC). Causes of death were COVID-19-related acute respiratory distress syndrome (UC:10, CCP:3), cardiac origin (UC:0, CCP:2), sepsis (CCP:2, UC:3), gastro-intestinal (CCP:0, UC:1), vascular (CCP:1, UC:0), and 1 of unknown origin (CCP). ## DISCUSSION In the current trial, CCP treatment failed to show a better efficacy than usual care for treating hospitalised COVID-19 patients not requiring assisted ventilation, although a trend in improved survival in the CCP arm was observed as early as d14 and confirmed at d28, without reaching a level of significance. A trend towards early pulmonary function worsening after CCP administration was evidenced. The absence of efficacy associated with CCP agrees with the results of most clinical trials currently reported in hospitalized COVID-19 patients.19 Indeed, only a limited number of randomized studies have reported a better survival following CCP treatment,20,21 while several other trials, notably the large RECOVERY trial,22 have found no evidence of survival benefit with CCP. Reasons for these discrepancies may relate to CCP characteristics, time to treatment from first symptoms, treatment modalities, and patient characteristics. Importantly, our study included a significant proportion of patients with underlying immunosuppression. In agreement with prior findings,23 such COVID-19 patients have a worse prognosis, as noted in the UC arm. In these patients with underlying immunosuppression, CCP treatment was associated with improved survival. Several studies have suggested that CCP may be particularly effective in patients unable to mount an immune response, notably a humoral response. We reported earlier that CCP treatment in immunosuppressed patients, mainly B-cell haematological malignancies treated by anti-CD-20 monoclonal antibodies, was associated with a favorable outcome.6 Further evidence was provided by two independent exposed/non-exposed studies with propensity score in patients with underlying immunosuppression. Hazard ratios of 0.52 [95%CI 0.29-0.92] and 0.50 [95%CI 0.34-0.72] were reported in favour of CCP treatment, respectively.6,7 Such reduction in mortality is strikingly close to the reduction in mortality observed in similar patients randomized to the CCP arm of the CORIPLASM trial. Most of the other randomised trials published to date did not report subgroup analysis for patients with underlying immunosuppression. One notable exception is the REMAP-CAP trial that investigated CCP in critically ill COVID-19 patients.22 Although the overall results of this study did not provide evidence of the efficacy of CCP in such patients, a pre-specified subgroup analysis revealed a potential benefit of CCP in patients with immunodeficiency. An antibody (Ab)-dose effect has been evidenced in several randomized studies3,24 as well as in the early access program in the United States.25 In the CORIPLASM study, 800-880 ml of CCP were transfused to patients randomised to the CCP arm. In most reported studies to date, patients most often received 250 ml-500 ml of CCP, with the notable exception of the CAPSID trial where CCP patients received 700-750 ml.24 Interestingly, the CAPSID trial reported a significant Ab-dose effect regarding several outcomes, including survival at d28. Differently from CAPSID, the CORIPLAM protocol recommended four CCP units provided by different donors for each patient, which resulted in less variation in mean Ab content in transfused CCP among included patients. This difference in transfusion practice may have contributed to reducing the ability to identify an Ab dose effect in our study. Furthermore, CCP for the CORIPLASM study were collected early in the COVD-19 crisis, when vaccination was not yet available, and prior to the occurrence of relevant SARS-CoV-2 variants. Several studies have demonstrated that plasma provided by convalescent vaccinated donors not only strongly increased anti-SARS-CoV-2 Ab titres and sero-neutralisation ratios but also increased cross-reactivity with a broader spectrum with respect to variants to which the donor has not been exposed.12,26 Plasma from such convalescent vaccinated donors may be endowed with increased clinical efficacy. Early intervention with CCP has been associated with improved outcome.3,4 Patients in our study exhibited a median of 7 days of symptoms at the time of inclusion, which is a short time period compared to most reported trials involving hospitalised patients. However, pre-specified subgroup analysis did not favour increased CCP efficacy associated with a shorter time period since symptoms onset. The high frequency of patients with underlying immunosuppression, for whom seroconversion is not expected early on, may contribute. Also, and as observed in other COVID-19 trials, early hospitalisation may be associated with more severe disease.8,22 Patients in the CCP arm tended to exhibit worsening pulmonary clinical conditions compared to patients in the UC arm early after transfusion. The occurrence of early transient pulmonary worsening after CCP transfusion has been reported elsewhere as well27, and may be in relation to an antibody-dependent enhancement involving immune-complex mediated inflammatory immunopathology in infected tissues.28 Also, an Ab-dependent FcR-mediated infection of tissues macrophages (and circulating monocytes) may result in a massive inflammatory response, as recently evidenced29, and may contribute as well. In fact, such early outcomes are seldomly reported in clinical studies. Furthermore, an early pulmonary worsening may be challenging to distinguish from transfusion associated circulatory overload (TACO), transfusion-related acute lung injury (TRALI) or overall disease worsening, possibly initiated before transfusion. Importantly such early worsening did not prevent subsequent improvement and a trend towards increased survival as early as d14 post randomisation. Of note, the observation that antibody-mediated SARS-CoV-2 uptake by monocytes/macrophages triggers inflammatory cell death and inhibition of viral replication may provide a mechanism for subsequent disease improvement.30 Our study has some limitations. The relatively small size of the trial limited the ability to appropriately assess outcomes such as patient mortality. Nevertheless, this did not prevent several important findings, notably regarding CCP treatment in immunosuppressed patients. Also, information regarding patient serostatus at inclusion was often unavailable, preventing meaningful findings in this regard. Overall, CCP treatment did not improve early outcome in patients with moderate-to-severe COVID-19 pneumonia. CCP-associated early respiratory worsening as well as CCP-associated reduced d14 and d28 mortality were observed, while not reaching statistical significance. CCP treatment was associated with reduced d28 mortality in patients with underlying immunosuppression. In addition, the recent emergence of the omicron variant with its BA.1 to BA.5 subvariants has highlighted the risks associated with immune-resistant SARS-CoV-2 and loss of efficacy of available monoclonal antibodies.11 While several months are necessary to produce one or more new monoclonal antibodies more suited to the evolution of circulating viral strains, convalescent plasma, notably from vaccinated donors, has demonstrated increased resilience to immune-resistant SARS-CoV-2 variants,12, 26 increased scalability as it may rely on existing collection infrastructure, as well as increased adaptability. The time between the onset of a COVID-19 variant and the availability of convalescent plasma from donors infected with the given variant disease is approximately four weeks. These results, along with the recent data obtained from other trials and cohort studies may support the use of convalescent plasma in immunocompromised patients for whom therapeutic options are currently scarce. ## Supporting information Appendix [[supplements/278329_file03.pdf]](pending:yes) ## Data Availability The data for this article will be made available after publication on request from any qualified researchers or academicians. The data include: analysed deidentified participant data, data dictionary, study protocol, statistical analysis plan, and the informed consent form, among other data. The data will be shared for 2 years after publication upon receipt of a request sent to raphael.porcher{at}aphp.fr. ## Role of the funding source The funders of the study had no role in study design, data collection, data analysis, data interpretation, or writing of the report. ## Contributors KL and PT were involved in the protocol design and study design, including conceptualization, methodology, funding acquisition, and resources. NS and TS did the data curation, investigation, project administration, supervision, and validation. RP and GB did the data analysis. TH, AM, TC, SGL, FA, JS, GMB, JMM, FB, CC, OH, VP, MM, XL, PB, FP, DS, SR, LR and JMM included the patients. OH, XM, PLT, MRR and PR designed the CORIMUNO19 platform trial and conceptualized the embedded trials. The manuscript was drafted by KL and PT and read and edited by all authors who approved the final manuscript. RP and GB had direct access to data. ## Declaration of interests PT, PM and AF are employees of Etablissement Français du Sang (EFS), the French public transfusion service, which collects, manufactures, tests and issues all blood components in France. All other co-authors declare no competing interests related to the topic of the study. ## Fundings Programme Hospitalier de recherche Clinique (DGOS-French Ministry of Health), Fondation pour la Recherche Médicale, Sorbonne Université AAP 2020, Emergency Support Instrument (ESI), DG Santé, European Commission. A CC-BY public copyright license (CC-BY 4.0 [https://creativecommons.org/licenses/by/4.0/](https://creativecommons.org/licenses/by/4.0/)) has been applied by the authors to the present document and will be applied to all subsequent versions up to the Author Accepted Manuscript arising from this submission, in accordance with the grant’s open access conditions. ## Data sharing The data for this article will be made available after publication on request from any qualified researchers or academicians. The data include: analysed deidentified participant data, data dictionary, study protocol, statistical analysis plan, and the informed consent form, among other data. The data will be shared for 2 years after publication upon receipt of a request sent to raphael.porcher{at}aphp.fr. ## APPENDIX CORIMUNO-19 Collaborative Group Supplementary Methods Supplementary Results ## Acknowledgements The authors wish to thank all physicians, nurses, and assistant nurses who took care of the patients, clinical research assistants and clinical research doctors who included and followed the patients during the trial, physicians, nurses and staff involved in convalescent plasma collection, manufacturing, testing and issuing, and above all the patients who agreed to participate in the study and the convalescent donors who generously gave their plasma. Special thanks to the DRCI of Assistance Publique-Hôpitaux de Paris (APHP), the trial sponsor, and the Unité de Recherche de l’Est Parisien (URC-EST, APHP.SU, site St Antoine site), which managed the trial. * Received August 9, 2022. * Revision received August 9, 2022. * Accepted August 10, 2022. * © 2022, Posted by Cold Spring Harbor Laboratory The copyright holder for this pre-print is the author. All rights reserved. The material may not be redistributed, re-used or adapted without the author's permission. ## REFERENCES 1. Casadevall A, Pirofski LA. The convalescent sera option for containing COVID-19. J Clin Invest. 2020 Apr 1;130(4):1545–1548. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 2. Piechotta V, Iannizzi C, Chai KL, et al. Convalescent plasma or hyperimmune immunoglobulin for people with COVID-19: a living systematic review. Cochrane Database Syst Rev 2021; 5: CD013600. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/14651858.CD013600.pub4&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 3. Libster R, Pérez Marc G, Wappner D, et al. Early High-Titer Plasma Therapy to Prevent Severe Covid-19 in Older Adults. N Engl J Med. 2021; 384: 610–618. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2033700&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 4. Sullivan DJ, Gebo KA, Shoham S, et al. Early Outpatient Treatment for Covid-19 with Convalescent Plasma. N Engl J Med 2022; 386:1700–1711. 5. Thompson MA, Henderson JP, Shah PK, et al. Association of Convalescent Plasma Therapy With Survival in Patients With Hematologic Cancers and COVID-19. JAMA Oncol. 2021; 7: 1167–75. 6. Hueso T, Godron AS, Lanoy E, et al. Convalescent plasma improves overall survival in patients with B-cell lymphoid malignancy and COVID-19: a longitudinal cohort and propensity score analysis. Leukemia. 2022; 36: 1025–1034. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/s41375-022-01511-6&link_type=DOI) 7. Weinreich DM, Sivapalasingam S, Norton T, et al. REGEN-COV Antibody Combination and Outcomes in Outpatients with Covid-19. N Engl J Med 2021; 385: e81. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/nejmoa2108163&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 8. RECOVERY Collaborative Group. Casirivimab and imdevimab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. Lancet 2022; 399: 665–676. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(22)00163-5&link_type=DOI) 9. Persad G, Peek ME, Shah SK. Fair Allocation of Scarce Therapies for COVID-19. Clin Infect Di. 2021; ciab1039. 10. Planas D, Veyer D, Baidaliuk A, et al. Reduced sensitivity of SARS-CoV-2 variant Delta to antibody neutralization. Nature. 2021; 596: 276–280. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1101/2021.05.26.445838&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 11. Planas D, Saunders N, Maes P, et al. Considerable escape of SARS-CoV-2 Omicron to antibody neutralization. Nature 2022; 602: 671–675. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 12. Li M, Beck EJ, Laeyendecker O, Eby Y, et al. Convalescent plasma with a high level of virus-specific antibody effectively neutralizes SARS-CoV-2 variants of concern. Blood Adv. 2022; 6 :3678–3683. 13. Rössler A, Riepler L, Bante D, von Laer D, Kimpel J. SARS-CoV-2 Omicron Variant Neutralization in Serum from Vaccinated and Convalescent Persons. N Engl J Med 2022; 386: 698–700. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=35021005&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 14. Jayk Bernal A, Gomes da Silva MM, Musungaie DB, et al. Molnupiravir for Oral Treatment of Covid-19 in Nonhospitalized Patients. N Engl J Med 2022; 386: 509–520. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 15. Hammond J, Leister-Tebbe H, Gardner A, et al. Oral Nirmatrelvir for High-Risk, Non hospitalized Adults with Covid-19. N Engl J Med 2022 Feb 16; 386:1397–1408 [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/nejmoa2118542&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 16. Beigel JH, Tomashek KM, Dodd LE, et al. Remdesivir for the Treatment of Covid-19 - Final Report. N Engl J Med. 2020; 383: 1813–1826. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2007764&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 17. Hermine O, Mariette X, Tharaux PL, et al. Effect of Tocilizumab vs Usual Care in Adults Hospitalized With COVID-19 and Moderate or Severe Pneumonia: A Randomized Clinical Trial. JAMA Intern Med 2021; 181: 32–40. 18. Gallian P, Pastorino B, Morel P, et al. Lower prevalence of antibodies neutralizing SARS-CoV-2 in group O French blood donors. Antiviral Res 2020; 181: 104880. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.antiviral.2020.104880&link_type=DOI) 19. Janiaud P, Axfors C, Schmitt AM, et al. Association of Convalescent Plasma Treatment With Clinical outcomes in Patients With COVID-19: A Systematic Review and Meta-analysis. JAMA 2021; 325: 1185–1195. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 20. O’Donnell MR, Grinsztejn B, Cummings MJ, et al. A randomized double-blind controlled trial of convalescent plasma in adults with severe COVID-19. J Clin Invest 2021; 131: e150646. 21. Bar KJ, Shaw PA, Choi GH, Aqui N, et al. A randomized controlled study of convalescent plasma for individuals hospitalized with COVID-19 pneumonia. J Clin Invest. 2021; 131: e155114. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI155114&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 22. Estcourt LJ, Turgeon AF, McQuilten, et al. Effect of Convalescent Plasma on Organ Support-Free Days in Critically Ill Patients With COVID-19: A Randomized Clinical Trial. JAMA 2021; 326: 1690–1702. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 23. Duléry R, Lamure S, Delord M, et al. Prolonged in-hospital stay and higher mortality after Covid-19 among patients with non-Hodgkin lymphoma treated with B-cell depleting immunotherapy. Am J Hematol 2021; 96: 934–944. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 24. Körper S, Weiss M, Zickler D, et al. Results of the CAPSID randomized trial for high-dose convalescent plasma in patients with severe COVID-19. J Clin Invest 2021; 131: e152264. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 25. Joyner MJ, Carter RE, Senefeld JW, et al. Convalescent Plasma Antibody Levels and the Risk of Death from Covid-19. N Engl J Med. 2021; 384: 1015–1027. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1056/NEJMoa2031893&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 26. Stamatatos L, Czartoski J, Wan YH, et al. mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection. Science 2021; 372: 1413–1418. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6Mzoic2NpIjtzOjU6InJlc2lkIjtzOjEzOiIzNzIvNjU0OS8xNDEzIjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjIvMDgvMTAvMjAyMi4wOC4wOS4yMjI3ODMyOS5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 27. Avendaño-Solá C, Ramos-Martínez A, Muñez-Rubio E, et al. A multicenter randomized open-label clinical trial for convalescent plasma in patients hospitalized with COVID-19 pneumonia. J Clin Invest. 2021; 131: e152740. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1172/JCI152740&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2022%2F08%2F10%2F2022.08.09.22278329.atom) 28. Lee WS, Wheatley AK, Kent SJ, DeKosky BJ. Antibody-dependent enhancement and SARS-CoV-2 vaccines and therapies. Nat Microbiol. 2020; 5: 1185–1191. 29. Junqueira C, Crespo Â, Ranjbar S, et al. FcγR-mediated SARS-CoV-2 infection of monocytes activates inflammation. Nature 2022; 606: 576–584. 30. Sefik E, Qu R, Junqueira C, Kaffe E, et al. Inflammasome activation in infected macrophages drives COVID-19 pathology. Nature. 2022; 606: 585–593.